101
|
Wright DFB, Duffull SB, Merriman TR, Dalbeth N, Barclay ML, Stamp LK. Predicting allopurinol response in patients with gout. Br J Clin Pharmacol 2015; 81:277-89. [PMID: 26451524 DOI: 10.1111/bcp.12799] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/25/2015] [Accepted: 10/03/2015] [Indexed: 12/11/2022] Open
Abstract
AIMS The primary aim of this research was to predict the allopurinol maintenance doses required to achieve the target plasma urate of ≤0.36 mmol l(-1) . METHODS A population analysis was conducted in nonmem using oxypurinol and urate plasma concentrations from 133 gout patients. Maintenance dose predictions to achieve the recommended plasma urate target were generated. RESULTS The urate response was best described by a direct effects model. Renal function, diuretic use and body size were found to be significant covariates. Dose requirements increased approximately 2-fold over a 3-fold range of total body weight and were 1.25-2 fold higher in those taking diuretics. Renal function had only a modest impact on dose requirements. CONCLUSIONS Contrary to current guidelines, the model predicted that allopurinol dose requirements were determined primarily by differences in body size and diuretic use. A revised guide to the likely allopurinol doses to achieve the target plasma urate concentration is proposed.
Collapse
Affiliation(s)
| | | | | | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland
| | - Murray L Barclay
- Department of Medicine, University of Otago, Christchurch.,Department of Clinical Pharmacology, Christchurch Hospital, Christchurch, New Zealand
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch
| |
Collapse
|
102
|
Nakajima A, Oda S, Yokoi T. Allopurinol induces innate immune responses through mitogen-activated protein kinase signaling pathways in HL-60 cells. J Appl Toxicol 2015; 36:1120-8. [PMID: 26641773 DOI: 10.1002/jat.3272] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/21/2015] [Accepted: 10/29/2015] [Indexed: 11/06/2022]
Abstract
Allopurinol, an inhibitor of xanthine oxidase, is a frequent cause of severe cutaneous adverse reactions (SCARs) in humans, including drug rash with eosinophilia and systemic symptoms, Stevens-Johnson syndrome and toxic epidermal necrolysis. Although SCARs have been suspected to be immune-mediated, the mechanisms of allopurinol-induced SCARs remain unclear. In this study, we examined whether allopurinol has the ability to induce innate immune responses in vitro using human dendritic cell (DC)-like cell lines, including HL-60, THP-1 and K562, and a human keratinocyte cell line, HaCaT. In this study, we demonstrate that treatment of HL-60 cells with allopurinol significantly increased the mRNA expression levels of interleukin-8, monocyte chemotactic protein-1 and tumor necrosis factor α in a time- and concentration-dependent manner. Furthermore, allopurinol induced the phosphorylation of mitogen-activated protein kinases (MAPK), such as c-Jun N-terminal kinase and extracellular signal-regulated kinase, which regulate cytokine production in DC. In addition, allopurinol-induced increases in cytokine expression were inhibited by co-treatment with the MAPK inhibitors. Collectively, these results suggest that allopurinol has the ability to induce innate immune responses in a DC-like cell line through activation of the MAPK signaling pathways. These results indicate that innate immune responses induced by allopurinol might be involved in the development of allopurinol-induced SCARs. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Akira Nakajima
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shingo Oda
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
103
|
Chung WH, Chang WC, Stocker SL, Juo CG, Graham GG, Lee MHH, Williams KM, Tian YC, Juan KC, Jan Wu YJ, Yang CH, Chang CJ, Lin YJ, Day RO, Hung SI. Insights into the poor prognosis of allopurinol-induced severe cutaneous adverse reactions: the impact of renal insufficiency, high plasma levels of oxypurinol and granulysin. Ann Rheum Dis 2015; 74:2157-64. [PMID: 25115449 DOI: 10.1136/annrheumdis-2014-205577] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 07/26/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Allopurinol, an antihyperuricaemic agent, is one of the common causes of life-threatening severe cutaneous adverse reactions (SCAR), including drug rash with eosinophilia and systemic symptoms (DRESS), Stevens-Johnson syndrome (SJS) and toxic epidermal necrosis (TEN). The prognostic factors for allopurinol-related SCAR remain unclear. This study aimed to investigate the relationship of dosing, renal function, plasma levels of oxypurinol and granulysin (a cytotoxic protein of SJS/TEN), the disease severity and mortality in allopurinol-SCAR. METHODS We prospectively enrolled 48 patients with allopurinol-SCAR (26 SJS/TEN and 22 DRESS) and 138 allopurinol-tolerant controls from 2007 to 2012. The human leucocyte antigen (HLA)-B*58:01 status, plasma concentrations of oxypurinol and granulysin were determined. RESULTS In this cohort, HLA-B*58:01 was strongly associated with allopurinol-SCAR (p<0.001, OR (95% CI) 109 (25 to 481)); however, the initial/maintenance dosages showed no relationship with the disease. Poor renal function was significantly associated with the delayed clearance of plasma oxypurinol, and increased the risk of allopurinol-SCAR (p<0.001, OR (95% CI) 8.0 (3.9 to 17)). Sustained high levels of oxypurinol after allopurinol withdrawal correlated with the poor prognosis of allopurinol-SCAR. In particular, the increased plasma levels of oxypurinol and granulysin linked to the high mortality of allopurinol-SJS/TEN (p<0.01), and strongly associated with prolonged cutaneous reactions in allopurinol-DRESS (p<0.05). CONCLUSIONS Impaired renal function and increased plasma levels of oxypurinol and granulysin correlated with the poor prognosis of allopurinol-SCAR. Allopurinol prescription is suggested to be avoided in subjects with renal insufficiency and HLA-B*58:01 carriers. An early intervention to increase the clearance of plasma oxypurinol may improve the prognosis of allopurinol-SCAR.
Collapse
Affiliation(s)
- Wen-Hung Chung
- Department of Dermatology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wan-Chun Chang
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Sophie L Stocker
- Department of Clinical Pharmacology & Toxicology, St. Vincent's Hospital, Sydney, New South Wales, Australia St Vincent's Hospital Clinical School and School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Chiun-Gung Juo
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Garry G Graham
- Department of Clinical Pharmacology & Toxicology, St. Vincent's Hospital, Sydney, New South Wales, Australia St Vincent's Hospital Clinical School and School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Ming-Han H Lee
- Department of Clinical Pharmacology & Toxicology, St. Vincent's Hospital, Sydney, New South Wales, Australia St Vincent's Hospital Clinical School and School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Kenneth M Williams
- Department of Clinical Pharmacology & Toxicology, St. Vincent's Hospital, Sydney, New South Wales, Australia St Vincent's Hospital Clinical School and School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Ya-Chung Tian
- College of Medicine, Chang Gung University, Taoyuan, Taiwan Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Kuo-Chang Juan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Yeong-Jian Jan Wu
- College of Medicine, Chang Gung University, Taoyuan, Taiwan Division of Allergy, Immunology and Rheumatology, Department of Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chih-Hsun Yang
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chee-Jen Chang
- Graduate Institute of Clinical Medical Science, Clinical Informatics and Medical Statistics Research Center, Chang Gung University, Taoyuan, Taiwan Biostatistical Center for Clinical Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-Jr Lin
- Graduate Institute of Clinical Medical Science, Clinical Informatics and Medical Statistics Research Center, Chang Gung University, Taoyuan, Taiwan Biostatistical Center for Clinical Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Richard O Day
- Department of Clinical Pharmacology & Toxicology, St. Vincent's Hospital, Sydney, New South Wales, Australia St Vincent's Hospital Clinical School and School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Shuen-Iu Hung
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
104
|
Stamp LK, Day RO, Yun J. Allopurinol hypersensitivity: investigating the cause and minimizing the risk. Nat Rev Rheumatol 2015; 12:235-42. [PMID: 26416594 DOI: 10.1038/nrrheum.2015.132] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Allopurinol is the most commonly prescribed urate-lowering therapy for the management of gout. Serious adverse reactions associated with allopurinol, while rare, are feared owing to the high mortality. Such reactions can manifest as a rash combined with eosinophilia, leukocytosis, fever, hepatitis and progressive kidney failure. Risk factors for allopurinol-related severe adverse reactions include the recent introduction of allopurinol, the presence of the HLA-B(*)58:01 allele, and factors that influence the drug concentration. The interactions between allopurinol, its metabolite, oxypurinol, and T cells have been studied, and evidence exists that the presence of the HLA-B(*)58:01 allele and a high concentration of oxypurinol function synergistically to increase the number of potentially immunogenic-peptide-oxypurinol-HLA-B(*)58:01 complexes on the cell surface, thereby increasing the risk of T-cell sensitization and a subsequent adverse reaction. This Review will discuss the above issues and place this in the clinical context of reducing the risk of serious adverse reactions.
Collapse
Affiliation(s)
- Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, P.O. Box 4345, Christchurch 8140, New Zealand
| | - Richard O Day
- Department of Clinical Pharmacology &Toxicology, St Vincent's Hospital, Darlinghurst, NSW 2010, Australia
| | - James Yun
- Department of Clinical Immunology and Allergy, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| |
Collapse
|
105
|
Perez-Ruiz F, Chinchilla SP, Atxotegi J, Urionagüena I, Herrero-Beites AM, Aniel-Quiroga MA. Increase in thyroid stimulating hormone levels in patients with gout treated with inhibitors of xanthine oxidoreductase. Rheumatol Int 2015; 35:1857-61. [PMID: 26342297 DOI: 10.1007/s00296-015-3355-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/26/2015] [Indexed: 10/23/2022]
Abstract
Increase in thyroid stimulating hormone (TSH) levels over the upper normal limit has been reported in a small percentage of patients treated with febuxostat in clinical trials, but a mechanistic explanation is not yet available. In an observational parallel longitudinal cohort study, we evaluated changes in TSH levels in patients with gout at baseline and during urate-lowering treatment with febuxostat. Patients to be started on allopurinol who had a measurement of TSH in the 6-month period prior to baseline evaluation were used for comparison. TSH levels and change in TSH levels at 12-month follow-up were compared between groups. Patients with abnormal TSH levels or previous thyroid disease or on amiodarone were not included for analysis. Eighty-eight patients treated with febuxostat and 87 with allopurinol were available for comparisons. Patients to be treated with febuxostat had higher urate levels and TSH levels, more severe gout, and poorer renal function, but were similar regarding other characteristics. A similar rise in TSH levels was observed in both groups (0.4 and 0.5 µUI/mL for febuxostat and allopurinol, respectively); at 12-mo, 7/88 (7.9 %) of patients on febuxostat and 4/87 (3.4 %) of patients on allopurinol showed TSH levels over 0.5 µUI/mL. Doses prescribed (corrected for estimated glomerular filtration rate in the case if patients on allopurinol) and baseline TSH levels were determinants of TSH levels at 12-month follow-up. No impact on free T4 (fT4) levels was observed. Febuxostat, but also allopurinol, increased TSH levels in a dose-dependent way, thus suggesting rather a class effect than a drug effect, but with no apparent impact on either clinical or fT4 levels.
Collapse
Affiliation(s)
- Fernando Perez-Ruiz
- Rheumatology Division, Hospital Universitario Cruces and Biocruces Health Research Institute, Pza. Cruces sn, 48903, Baracaldo, Spain.
| | | | - Joana Atxotegi
- Rheumatology Division, Hospital Universitario Cruces and Biocruces Health Research Institute, Pza. Cruces sn, 48903, Baracaldo, Spain
| | - Irati Urionagüena
- Rheumatology Division, Hospital Universitario Cruces, Baracaldo, Spain
| | - Ana Maria Herrero-Beites
- Physical Medicine Division, Hospital de Górliz and Biocruces Helath Research Institute, Górliz, Spain
| | | |
Collapse
|
106
|
Givertz MM, Anstrom KJ, Redfield MM, Deswal A, Haddad H, Butler J, Tang WHW, Dunlap ME, LeWinter MM, Mann DL, Felker GM, O'Connor CM, Goldsmith SR, Ofili EO, Saltzberg MT, Margulies KB, Cappola TP, Konstam MA, Semigran MJ, McNulty SE, Lee KL, Shah MR, Hernandez AF. Effects of Xanthine Oxidase Inhibition in Hyperuricemic Heart Failure Patients: The Xanthine Oxidase Inhibition for Hyperuricemic Heart Failure Patients (EXACT-HF) Study. Circulation 2015; 131:1763-71. [PMID: 25986447 PMCID: PMC4438785 DOI: 10.1161/circulationaha.114.014536] [Citation(s) in RCA: 231] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 03/05/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND Oxidative stress may contribute to heart failure (HF) progression. Inhibiting xanthine oxidase in hyperuricemic HF patients may improve outcomes. METHODS AND RESULTS We randomly assigned 253 patients with symptomatic HF, left ventricular ejection fraction ≤40%, and serum uric acid levels ≥9.5 mg/dL to receive allopurinol (target dose, 600 mg daily) or placebo in a double-blind, multicenter trial. The primary composite end point at 24 weeks was based on survival, worsening HF, and patient global assessment. Secondary end points included change in quality of life, submaximal exercise capacity, and left ventricular ejection fraction. Uric acid levels were significantly reduced with allopurinol in comparison with placebo (treatment difference, -4.2 [-4.9, -3.5] mg/dL and -3.5 [-4.2, -2.7] mg/dL at 12 and 24 weeks, respectively, both P<0.0001). At 24 weeks, there was no significant difference in clinical status between the allopurinol- and placebo-treated patients (worsened 45% versus 46%, unchanged 42% versus 34%, improved 13% versus 19%, respectively; P=0.68). At 12 and 24 weeks, there was no significant difference in change in Kansas City Cardiomyopathy Questionnaire scores or 6-minute walk distances between the 2 groups. At 24 weeks, left ventricular ejection fraction did not change in either group or between groups. Rash occurred more frequently with allopurinol (10% versus 2%, P=0.01), but there was no difference in serious adverse event rates between the groups (20% versus 15%, P=0.36). CONCLUSIONS In high-risk HF patients with reduced ejection fraction and elevated uric acid levels, xanthine oxidase inhibition with allopurinol failed to improve clinical status, exercise capacity, quality of life, or left ventricular ejection fraction at 24 weeks. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00987415.
Collapse
Affiliation(s)
- Michael M Givertz
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.).
| | - Kevin J Anstrom
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Margaret M Redfield
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Anita Deswal
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Haissam Haddad
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Javed Butler
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - W H Wilson Tang
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Mark E Dunlap
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Martin M LeWinter
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Douglas L Mann
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - G Michael Felker
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Christopher M O'Connor
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Steven R Goldsmith
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Elizabeth O Ofili
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Mitchell T Saltzberg
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Kenneth B Margulies
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Thomas P Cappola
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Marvin A Konstam
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Marc J Semigran
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Steven E McNulty
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Kerry L Lee
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Monica R Shah
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| | - Adrian F Hernandez
- From Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.M.G.); Duke University Medical Center, Durham, NC (K.J.A., G.M.F., C.M.O., S.E.M., K.L.L., A.F.H.); Mayo Clinic, Rochester, MN (M.M.R.); Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, TX (A.D.); Ottawa Heart Institute, Ontario, Canada (H.H.); Emory University, Atlanta, GA (J.B.); Cleveland Clinic, OH (W.H.W.T.); MetroHealth Campus of Case Western Reserve University, Cleveland, OH (M.E.D.); University of Vermont, Burlington (M.M.L.); Washington University, St. Louis, MO (D.L.M.); Hennepin County Medical Center, Minneapolis, MN (S.R.G.); Morehouse School of Medicine, Atlanta, GA (E.O.O.); Christiana Care Health System, Newark, DE (M.T.S.); University of Pennsylvania, Philadelphia (K.B.M., T.P.C.); Tufts Medical Center, Boston, MA (M.A.K.); Massachusetts General Hospital, Boston (M.J.S.); and National Heart, Lung, and Blood Institute, Bethesda, MD (M.R.S.)
| |
Collapse
|
107
|
Funke J, Prasse C, Lütke Eversloh C, Ternes TA. Oxypurinol - A novel marker for wastewater contamination of the aquatic environment. WATER RESEARCH 2015; 74:257-265. [PMID: 25753675 DOI: 10.1016/j.watres.2015.02.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 06/04/2023]
Abstract
The anti-gout agent allopurinol is one of the most prescribed pharmaceuticals in Germany and is widely metabolized into oxypurinol (80%) as well as the corresponding riboside conjugates (10%) within the human body. To investigate the occurrence of allopurinol and oxypurinol in the urban water cycle an analytical method was developed based on solid phase extraction (SPE) and subsequent liquid chromatography electrospray-ionization tandem mass spectrometry (LC-MS/MS). In raw wastewater concentration levels of oxypurinol ranged up to 26.6 μg L(-1), whereas allopurinol was not detected at all. In wastewater treatment plant (WWTP) effluents, concentrations of allopurinol were <LOQ, whereas oxypurinol concentrations ranged from 2.3 μg L(-1) to 21.7 μg L(-1). Elevated concentrations of oxypurinol in biologically treated wastewater originate from the transformation of allopurinol as well as the cleavage of allopurinol-9-riboside, which was confirmed by laboratory experiments with activated sludge taken from a municipal WWTP. Further tracking of oxypurinol in the urban water cycle revealed its presence in rivers and streams (up to 22.6 μg L(-1)), groundwater (up to 0.38 μg L(-1)) as well as in finished drinking water (up to 0.30 μg L(-1)). Due to the high biological stability and the almost ubiquitous presence in the urban water cycle at elevated concentrations, oxypurinol might be used as marker for domestic wastewater in the environment. This was confirmed by correlation analysis to other wastewater markers with strong correlations of the concentrations of oxypurinol and carbamazepine (r(2) = 0.89) as well as of oxypurinol and primidone (r(2) = 0.82).
Collapse
Affiliation(s)
- Jan Funke
- Federal Institute of Hydrology (BfG), Am Mainzer Tor 1, 56068 Koblenz, Germany
| | - Carsten Prasse
- Federal Institute of Hydrology (BfG), Am Mainzer Tor 1, 56068 Koblenz, Germany
| | | | - Thomas A Ternes
- Federal Institute of Hydrology (BfG), Am Mainzer Tor 1, 56068 Koblenz, Germany.
| |
Collapse
|
108
|
Lu G, Goodman AM, Ayres B, Builta Z, Haes AJ. Near real-time determination of metabolic parameters for unquenched 6-mercaptopurine and xanthine oxidase samples using capillary electrophoresis. J Pharm Biomed Anal 2015; 111:51-6. [PMID: 25863016 DOI: 10.1016/j.jpba.2015.03.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/16/2015] [Accepted: 03/17/2015] [Indexed: 11/29/2022]
Abstract
The enzyme activity of xanthine oxidase (XO) is influenced by several environmental factors including solution conditions, storage conditions, inhibitors, other enzymes, and activators. For instance, the metabolic reaction involving XO and the pro-drug 6-mercaptopurine, a drug used in the treatment maintenance of acute lymphatic leukemia, Crohn's disease, and ulcerative colitis, is often modified through the use of inhibitors, which varies the kinetic parameters associated with this reaction. Methods that provide fast and accurate determination of these kinetic constants can help in understanding the mechanism of these reactions. Herein, sequential and time-delayed electrokinetic injections of unpurified and unquenched samples containing xanthine oxidase, 6-mercaptopurine, and the inhibitor allopurinol are evaluated using capillary electrophoresis (CE). Using progress curve analysis, the Michaelis constant, apparent Michaelis constant, and inhibition constant are estimated to be 43.8 ± 2.0 μM, 143.0 ± 3.7 μM and 13.2 ± 1.4 μM, respectively. In addition, a turnover number of 7.9 ± 0.2 min(-1) is quantified. These values are consistent with some previously published values but were obtained without user intervention for reaction monitoring. This unique application of CE enzyme assays offers substantial advantages over traditional methods by determining kinetic parameters for enzymatic reactions with minimal (nL) sample volumes, short (<30 min) reaction analysis times, without any sample quenching or purification, and minimal user intervention.
Collapse
Affiliation(s)
- Grace Lu
- University of Iowa, Department of Chemistry, 204 IATL, Iowa City, IA 52242, United States
| | - Amanda M Goodman
- University of Iowa, Department of Chemistry, 204 IATL, Iowa City, IA 52242, United States; Rice University, Department of Chemistry, 6100 Main Street, Houston, TX 77005, United States
| | - Brennan Ayres
- University of Iowa, Department of Chemistry, 204 IATL, Iowa City, IA 52242, United States
| | - Zac Builta
- University of Iowa, Department of Chemistry, 204 IATL, Iowa City, IA 52242, United States
| | - Amanda J Haes
- University of Iowa, Department of Chemistry, 204 IATL, Iowa City, IA 52242, United States.
| |
Collapse
|
109
|
Sahoo S, Haraldsdóttir HS, Fleming RMT, Thiele I. Modeling the effects of commonly used drugs on human metabolism. FEBS J 2014; 282:297-317. [PMID: 25345908 DOI: 10.1111/febs.13128] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 09/21/2014] [Accepted: 10/13/2014] [Indexed: 02/02/2023]
Abstract
Metabolism contributes significantly to the pharmacokinetics and pharmacodynamics of a drug. In addition, diet and genetics have a profound effect on cellular metabolism with respect to both health and disease. In the present study, we assembled a comprehensive, literature-based drug metabolic reconstruction of the 18 most highly prescribed drug groups, including statins, anti-hypertensives, immunosuppressants and analgesics. This reconstruction captures in detail our current understanding of their absorption, intracellular distribution, metabolism and elimination. We combined this drug module with the most comprehensive reconstruction of human metabolism, Recon 2, yielding Recon2_DM1796, which accounts for 2803 metabolites and 8161 reactions. By defining 50 specific drug objectives that captured the overall drug metabolism of these compounds, we investigated the effects of dietary composition and inherited metabolic disorders on drug metabolism and drug-drug interactions. Our main findings include: (a) a shift in dietary patterns significantly affects statins and acetaminophen metabolism; (b) disturbed statin metabolism contributes to the clinical phenotype of mitochondrial energy disorders; and (c) the interaction between statins and cyclosporine can be explained by several common metabolic and transport pathways other than the previously established CYP3A4 connection. This work holds the potential for studying adverse drug reactions and designing patient-specific therapies.
Collapse
Affiliation(s)
- Swagatika Sahoo
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | | | | | | |
Collapse
|
110
|
Pavlos R, Mallal S, Ostrov D, Buus S, Metushi I, Peters B, Phillips E. T cell-mediated hypersensitivity reactions to drugs. Annu Rev Med 2014; 66:439-54. [PMID: 25386935 DOI: 10.1146/annurev-med-050913-022745] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The immunological mechanisms driving delayed hypersensitivity reactions (HSRs) to drugs mediated by drug-reactive T lymphocytes are exemplified by several key examples and their human leukocyte antigen (HLA) associations: abacavir and HLA-B*57:01, carbamazepine and HLA-B*15:02, allo-purinol and HLA-B*58:01, and both amoxicillin-clavulanate and nevirapine with multiple class I and II alleles. For HLA-restricted drug HSRs, specific class I and/or II HLA alleles are necessary but not sufficient for tissue specificity and the clinical syndrome. Several models have been proposed to explain the immunopathogenesis of severe T cell-mediated drug HSRs, and our increased understanding of the risk factors and mechanisms involved in the development of these reactions will further the development of sensitive and specific strategies for preclinical screening that will lead to safer and more cost-effective drug design.
Collapse
Affiliation(s)
- Rebecca Pavlos
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, 6150;
| | | | | | | | | | | | | |
Collapse
|
111
|
Helmy SA, El-Bedaiwy HM. Pharmacokinetics and comparative bioavailability of allopurinol formulations in healthy subjects. Clin Pharmacol Drug Dev 2014; 3:353-7. [PMID: 27129007 DOI: 10.1002/cpdd.95] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 11/20/2013] [Indexed: 11/12/2022]
Abstract
Allopurinol is the most commonly used urate-lowering therapy in gout. This study was undertaken to evaluate the pharmacokinetics and relative bioavailability of two brands of allopurinol tablets. The in vivo study was established according to a single-center, randomized, single-dose, laboratory-blinded, Two Way, Cross-Over Study with a washout period of 1 week. Under fasting conditions, 24 healthy male volunteers were randomly allocated to receive a single oral dose (200 mg) of either test and reference formulations. Plasma samples were obtained over a 6-hour interval and analyzed for allopurinol by reversed phase liquid chromatography with ultraviolet detection. The 90% confidence intervals for the ratio of log transformed values of Cmax , AUC0-t , and AUCt-∞ of the two treatments were within the acceptable range (0.8-1.25) for bioequivalence. From PK perspectives, the two allopurinol formulations were considered bioequivalent, based on the rate and extent of absorption. No adverse events occurred or were reported after a single 200-mg allopurinol and both formulations were well tolerated.
Collapse
Affiliation(s)
- Sally A Helmy
- Faculty of Pharmacy, Department of Pharmaceutics, Damanhour University, Damanhour, Egypt
| | - Heba M El-Bedaiwy
- Faculty of Pharmacy, Department of Industrial Pharmacy, Damanhour University, Damanhour, Egypt
| |
Collapse
|
112
|
Graham GG, Kannangara DRW, Stocker SL, Portek I, Pile KD, Indraratna PL, Datta I, Williams KM, Day RO. Understanding the dose-response relationship of allopurinol: predicting the optimal dosage. Br J Clin Pharmacol 2014; 76:932-8. [PMID: 23590252 DOI: 10.1111/bcp.12126] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 03/18/2013] [Indexed: 11/27/2022] Open
Abstract
AIMS The aim of the study was to identify and quantify factors that control the plasma concentrations of urate during allopurinol treatment and to predict optimal doses of allopurinol. METHODS Plasma concentrations of urate and creatinine (112 samples, 46 patients) before and during treatment with various doses of allopurinol (50-600 mg daily) were monitored. Non-linear and multiple linear regression equations were used to examine the relationships between allopurinol dose (D), creatinine clearance (CLcr) and plasma concentrations of urate before (UP) and during treatment with allopurinol (UT). RESULTS Plasma concentrations of urate achieved during allopurinol therapy were dependent on the daily dose of allopurinol and the plasma concentration of urate pre-treatment. The non-linear equation: UT = (1 - D/(ID50 + D)) × (UP - UR) + UR , fitted the data well (r(2) = 0.74, P < 0.0001). The parameters and their best fit values were: daily dose of allopurinol reducing the inhibitable plasma urate by 50% (ID50 = 226 mg, 95% CI 167, 303 mg), apparent resistant plasma urate (UR = 0.20 mmol l(-1), 95 % CI 0.14, 0.25 mmol l(-1)). Incorporation of CLcr did not significantly improve the fit (P = 0.09). CONCLUSIONS A high baseline plasma urate concentration requires a high dose of allopurinol to reduce plasma urate below recommended concentrations. This dose is dependent on only the pre-treatment plasma urate concentration and is not influenced by CLcr .
Collapse
Affiliation(s)
- Garry G Graham
- Department of Clinical Pharmacology & Toxicology, St Vincent's Hospital, NSW, Australia; School of Medical Sciences, University of New South Wales, NSW, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Stamp LK, Turner R, Khalilova IS, Zhang M, Drake J, Forbes LV, Kettle AJ. Myeloperoxidase and oxidation of uric acid in gout: implications for the clinical consequences of hyperuricaemia. Rheumatology (Oxford) 2014; 53:1958-65. [PMID: 24899662 DOI: 10.1093/rheumatology/keu218] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES The aims of this study were to establish whether, in patients with gout, MPO is released from neutrophils and urate is oxidized to allantoin and if these effects are attenuated by allopurinol. METHODS MPO, urate, allantoin and oxypurinol were measured in plasma from 54 patients with gout and 27 healthy controls. Twenty-three patients had acute gout, 13 of whom were receiving allopurinol, and 31 had intercritical gout, 20 of whom were receiving allopurinol. Ten additional gout patients had samples collected before and after 4 weeks of allopurinol. RESULTS Plasma MPO and its specific activity were higher (P < 0.05) in patients with acute gout not receiving allopurinol compared with controls. MPO protein in patients' plasma was related to urate concentration (r = 0.5, P < 0.001). Plasma allantoin was higher (P < 0.001) in all patient groups compared with controls. In controls and patients not receiving allopurinol, allantoin was associated with plasma urate (r = 0.62, P < 0.001) and MPO activity (r = 0.45, P < 0.002). When 10 patients were treated with allopurinol, it lowered their plasma urate and allantoin (P = 0.002). In all patients receiving allopurinol, plasma allantoin was related to oxypurinol (r = 0.65, P < 0.0001). Oxypurinol was a substrate for purified MPO that enhanced the oxidation of urate. CONCLUSION Increased concentrations of urate in gout lead to the release of MPO from neutrophils and the oxidation of urate. Products of MPO and reactive metabolites of urate may contribute to the pathology of gout and hyperuricaemia. At low concentrations, oxypurinol should reduce inflammation, but high concentrations may contribute to oxidative stress.
Collapse
Affiliation(s)
- Lisa K Stamp
- Department of Medicine, Department of Pathology, Centre for Free Radical Research, University of Otago, and Clinical Pharmacology Canterbury Health Laboratories, Christchurch, New Zealand
| | - Rufus Turner
- Department of Medicine, Department of Pathology, Centre for Free Radical Research, University of Otago, and Clinical Pharmacology Canterbury Health Laboratories, Christchurch, New Zealand
| | - Irada S Khalilova
- Department of Medicine, Department of Pathology, Centre for Free Radical Research, University of Otago, and Clinical Pharmacology Canterbury Health Laboratories, Christchurch, New Zealand
| | - Mei Zhang
- Department of Medicine, Department of Pathology, Centre for Free Radical Research, University of Otago, and Clinical Pharmacology Canterbury Health Laboratories, Christchurch, New Zealand
| | - Jill Drake
- Department of Medicine, Department of Pathology, Centre for Free Radical Research, University of Otago, and Clinical Pharmacology Canterbury Health Laboratories, Christchurch, New Zealand
| | - Louisa V Forbes
- Department of Medicine, Department of Pathology, Centre for Free Radical Research, University of Otago, and Clinical Pharmacology Canterbury Health Laboratories, Christchurch, New Zealand
| | - Anthony J Kettle
- Department of Medicine, Department of Pathology, Centre for Free Radical Research, University of Otago, and Clinical Pharmacology Canterbury Health Laboratories, Christchurch, New Zealand.
| |
Collapse
|
114
|
Hatoum H, Khanna D, Lin SJ, Akhras KS, Shiozawa A, Khanna P. Achieving serum urate goal: a comparative effectiveness study between allopurinol and febuxostat. Postgrad Med 2014; 126:65-75. [PMID: 24685969 DOI: 10.3810/pgm.2014.03.2741] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Febuxostat is recommended as 1 of 2 first-line urate-lowering therapies (ULT) for treating gout in the 2012 American College of Rheumatology Guidelines. Several efficacy trials have compared febuxostat with allopurinol treatment, but real-world comparative data are limited. METHODS We compared effectiveness of the 2 agents in reaching serum urate (sUA) level goal (< 6 mg/dL) within 6 months (main endpoint), factors impacting the likelihood of reaching goal, and outcomes in allopurinol patients who were switched to febuxostat therapy after failing to reach sUA level goal. Data from the General Electric Electronic Medical Record database on adult patients with newly diagnosed gout, who had started treatment with allopurinol or febuxostat in 2009 or thereafter were analyzed. Descriptive statistics, bivariate analyses, and logistic regressions were used. RESULTS Allopurinol (n = 17 199) and febuxostat (n = 1190) patients had a mean ± standard deviation (SD) age of 63.7 (± 13.37) years; most patients were men and white. Average daily medication doses (mg) in the first 6 months were 184.9 ± 96.7 and 48.4 ± 15.8 for allopurinol- and febuxostat-treated patients, respectively; 4.8% of allopurinol-treated patients switched to febuxostat, whereas 25.7% of febuxostat-treated patients switched to allopurinol. Febuxostat patients had lower estimated glomerular filtration rate levels, more diabetes mellitus, or tophi at baseline (P < 0.05) and 29.2% and 42.2% of patients in the allopurinol and febuxostat groups achieved goal sUA levels (P < 0.0001). Febuxostat was significantly more effective in patients reaching sUA goal (adjusted odds ratio, 1.73; 95% CI, 1.48-2.01). Older patients and women had greater likelihood of reaching sUA goal level; however, patients with higher Charlson Comorbidity Index scores, blacks, or those with estimated glomerular filtration rates between 15 to ≤ 60 mL/min had reduced likelihood of attaining goal (P < 0.05). Among allopurinol-treated patients who were switched to febuxostat after failing to reach goal, 244 (48.3%) reached goal on febuxostat (median = 62.5 days), with an average 39% sUA level reduction achieved within 6 months. Patients who did not reach goal had a 14.3% sUA level reduction. CONCLUSIONS The real-life data support the effectiveness of febuxostat in managing patients with gout.
Collapse
Affiliation(s)
- Hind Hatoum
- President, Hind T. Hatoum and Company; Adjunct Faculty, University of Illinois at Chicago, Chicago, IL
| | | | | | | | | | | |
Collapse
|
115
|
Richette P, Frazier A, Bardin T. Pharmacokinetics considerations for gout treatments. Expert Opin Drug Metab Toxicol 2014; 10:949-57. [PMID: 24809930 DOI: 10.1517/17425255.2014.915027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Patients with gout often have comorbid conditions such as renal failure, cardiovascular disease and metabolic syndrome. The presence and required treatment of these conditions can make the treatment of gout challenging. Knowledge of the pharmacokinetics of the available drugs for the management of gout is mandatory. AREAS COVERED A MEDLINE PubMed search for articles published in English from January 1990 to January 2014 was completed using the terms: pharmacokinetics, colchicine, canakinumab, allopurinol, febuxostat, pegloticase, gout, toxicity, drug interaction. EXPERT OPINION Colchicine is a drug with a narrow therapeutic-toxicity window. Co-prescription with strong CYP3A4 or P-glycoprotein inhibitors can greatly modify its pharmacokinetics and is to be avoided. Elimination of canakinumab mainly occurs via intracellular catabolism, following receptor mediator endocytosis. Canakinumab appears to be a good alternative for patients with contraindications to colchicine, NSAIDs and corticosteroids. For patients with renal impairment, some authors recommend that the allopurinol maximum dosage should be adjusted to creatinine clearance. If the urate target cannot be achieved, the therapy should be switched to febuxostat, which is appropriate with mild-to-moderate renal failure. Anti-pegloticase antibodies affect the pharmacokinetics of the drug because they increase its clearance, with loss of pegloticase activity.
Collapse
Affiliation(s)
- Pascal Richette
- Université Paris Diderot, Sorbonne Paris Cité, UFR de Médecine , F-75205 Paris , France
| | | | | |
Collapse
|
116
|
|
117
|
Sofue T, Inui M, Hara T, Nishijima Y, Moriwaki K, Hayashida Y, Ueda N, Nishiyama A, Kakehi Y, Kohno M. Efficacy and safety of febuxostat in the treatment of hyperuricemia in stable kidney transplant recipients. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:245-53. [PMID: 24600205 PMCID: PMC3933431 DOI: 10.2147/dddt.s56597] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Post-transplant hyperuricemia (PTHU), defined as serum uric acid concentration ≥7.0 mg/dL or need for treatment with allopurinol or benzbromarone, reduces long-term allograft survival in kidney transplant recipients. Febuxostat, a new nonpurine selective xanthine oxidase inhibitor, is well tolerated in patients with moderate renal impairment. However, its efficacy and safety in kidney recipients with PTHU is unclear. We therefore assessed the efficacy and safety of febuxostat in stable kidney transplant recipients with PTHU. Methods Of 93 stable adult kidney transplant recipients, 51 were diagnosed with PTHU (PTHU group) and 42 were not (NPTHU group). Of the 51 patients with PTHU, 26 were treated with febuxostat (FX group) and 25 were not (NFX group), at the discretion of each attending physician. One-year changes in serum uric acid concentrations, rates of achievement of target uric acid (<6.0 mg/dL), estimated glomerular filtration rates in allografts, and adverse events were retrospectively analyzed in the FX, NFX, and NPTHU groups. Results The FX group showed significantly greater decreases in serum uric acid (−2.0±1.1 mg/dL versus 0.0±0.8 mg/dL per year, P<0.01) and tended to show a higher rate of achieving target uric acid levels (50% versus 24%; odds ratio 3.17 [95% confidence interval 0.96–10.5], P=0.08) than the NFX group. Although baseline allograft estimated glomerular filtration rates tended to be lower in the FX group than in the NFX group (40±14 mL/min/1.73 m2 versus 47±19 mL/min/1.73 m2), changes in allograft estimated glomerular filtration rate were similar (+1.0±4.9 mL/min/1.73 m2 versus −0.2±6.9 mL/min/1.73 m2 per year, P=0.50). None of the patients in the FX group experienced any severe adverse effects, such as pancytopenia or attacks of gout, throughout the entire study period. Nephrologists were more likely than urologists to start febuxostat in kidney transplant recipients with PTHU (69% versus 8%). Conclusion Treatment with febuxostat sufficiently lowered uric acid levels without severe adverse effects in stable kidney transplant recipients with PTHU.
Collapse
Affiliation(s)
- Tadashi Sofue
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa
| | - Masashi Inui
- Department of Urology, Tokyo Women's Medical University, Tokyo
| | - Taiga Hara
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa
| | - Yoko Nishijima
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa
| | - Kumiko Moriwaki
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa
| | | | - Nobufumi Ueda
- Department of Urology, Kagawa University, Kagawa, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | | | - Masakazu Kohno
- Division of Nephrology and Dialysis, Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, Kagawa
| |
Collapse
|
118
|
An audit of a therapeutic drug monitoring service for allopurinol therapy. Ther Drug Monit 2013; 35:863-6. [PMID: 24263644 DOI: 10.1097/ftd.0b013e318299920a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Oxypurinol, the active metabolite of allopurinol, is the major determinant of the hypouricemic effect of allopurinol. Monitoring oxypurinol concentrations is undertaken to determine adherence to therapy, to investigate reasons for continuing attacks of acute gout and/or insufficiently low plasma urate concentrations despite allopurinol treatment, and to assess the risk of allopurinol hypersensitivity, an adverse effect that has been putatively associated with elevated plasma oxypurinol concentrations. METHODS An audit of request forms requesting plasma oxypurinol concentration measurements received by the pathology service (SydPath) at St Vincent's Hospital, Darlinghurst, Sydney was undertaken for the 7-year period January 2005-December 2011. Patient demographics, biochemical data, including plasma creatinine and uric acid concentrations, comorbidities, and concomitant medications were recorded. RESULTS There were 412 requests for determination of an oxypurinol concentration. On 48% of occasions, the time of allopurinol dosing was recorded, while just 79 (19%) blood samples were collected 6-9 hours postdosing, the time window used to establish the therapeutic range for oxypurinol. For these optimally interpretable concentrations, 32 (8%) were within the putative therapeutic range (5-15 mg/L), while 5 (1%) were below and 41 (10%) above this range. The daily dose of allopurinol was documented on only one-third of the request forms. Individually, plasma urate and creatinine concentrations were requested concomitantly with plasma oxypurinol concentrations in 66% and 58% of the cases, respectively; while plasma oxypurinol, urate, and creatinine concentrations were requested concomitantly in 49% of the cases. CONCLUSIONS Requesting clinicians and blood specimen collectors often fail to provide relevant information (dose, times of last dose, and blood sample collection) to allow the most useful interpretation of oxypurinol concentrations. Concomitant plasma urate and creatinine concentrations should be requested to allow more complete interpretation of the data.
Collapse
|
119
|
Liu X, Ni XJ, Shang DW, Zhang M, Hu JQ, Qiu C, Luo FT, Wen YG. Determination of allopurinol and oxypurinol in human plasma and urine by liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 941:10-6. [PMID: 24184830 DOI: 10.1016/j.jchromb.2013.09.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/17/2013] [Accepted: 09/19/2013] [Indexed: 11/28/2022]
Abstract
Allopurinol is used widely for the treatment of gout, but its pharmacokinetics is complex and some patients show hypersensitivity, necessitating careful monitoring and improved detection methods. In this study, a sensitive and reliable liquid chromatography-tandem mass spectrometry method was developed to determine the concentrations of allopurinol and its active metabolite oxypurinol in human plasma and urine using 2,6-dichloropurine as the internal standard (IS). Analytes and the IS were extracted from 0.5ml aliquots of plasma or urine using ethyl acetate and separated on an Agilent Eclipse Plus C18 column using methanol and ammonium formate-formic acid buffer containing 5mM ammonium formate and 0.1% formic acid (95:5, v/v) as the mobile phase (A) for allopurinol or methanol plus 5mM ammonium formate aqueous solution (95:5, v/v) as the mobile phase (B) for oxypurinol. Allopurinol was detected in positive ion mode and the analysis time was about 7min. The calibration curve was linear from 0.05 to 5μg/mL allopurinol in plasma and 0.5-30μg/mL in urine. The lower limit of quantification (LLOQ) was 0.05μg/mL in plasma and 0.5μg/mL in urine. The intra- and inter-day precision and relative errors of quality control (QC) samples were ≤11.1% for plasma and ≤ 8.7% for urine. Oxypurinol was detected in negative mode with an analysis time of about 4min. The calibration curve was linear from 0.05 to 5μg/mL in plasma (LLOQ, 0.05μg/mL) and from 1 to 50μg/mL in urine (LLOQ, 1μg/mL). The intra- and inter-day precision and relative errors were ≤7.0% for plasma and ≤9.6% for urine. This method was then successfully applied to investigate the pharmacokinetics of allopurinol and oxypurinol in humans.
Collapse
Affiliation(s)
- Xia Liu
- Department of Pharmacy, Guangzhou Brain Hospital, Guangzhou Medical University, 36 MingXin Road, Guangzhou 510370, China
| | | | | | | | | | | | | | | |
Collapse
|
120
|
Givertz MM, Mann DL, Lee KL, Ibarra JC, Velazquez EJ, Hernandez AF, Mascette AM, Braunwald E. Xanthine oxidase inhibition for hyperuricemic heart failure patients: design and rationale of the EXACT-HF study. Circ Heart Fail 2013; 6:862-8. [PMID: 23861505 DOI: 10.1161/circheartfailure.113.000394] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Michael M Givertz
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Ramasamy SN, Korb-Wells CS, Kannangara DRW, Smith MWH, Wang N, Roberts DM, Graham GG, Williams KM, Day RO. Allopurinol Hypersensitivity: A Systematic Review of All Published Cases, 1950–2012. Drug Saf 2013; 36:953-80. [DOI: 10.1007/s40264-013-0084-0] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
122
|
Vaughan VC, Martin P, Lewandowski PA. Cancer cachexia: impact, mechanisms and emerging treatments. J Cachexia Sarcopenia Muscle 2013; 4:95-109. [PMID: 23097000 PMCID: PMC3684701 DOI: 10.1007/s13539-012-0087-1] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 09/03/2012] [Indexed: 12/18/2022] Open
Abstract
Many forms of cancer present with a complex metabolic profile characterised by loss of lean body mass known as cancer cachexia. The physical impact of cachexia contributes to decreased patient quality of life, treatment success and survival due to gross alterations in protein metabolism, increased oxidative stress and systemic inflammation. The psychological impact also contributes to decreased quality of life for both patients and their families. Combination therapies that target multiple pathways, such as eicosapentaenoic acid administered in combination with exercise, appetite stimulants, antioxidants or anti-inflammatories, have potential in the treatment of this complex syndrome and require further development.
Collapse
Affiliation(s)
- Vanessa C Vaughan
- School of Medicine, Deakin University, 75 Pigdons Road, Waurn Ponds, Victoria, 3216, Australia
| | | | | |
Collapse
|
123
|
Obach RS. Pharmacologically active drug metabolites: impact on drug discovery and pharmacotherapy. Pharmacol Rev 2013; 65:578-640. [PMID: 23406671 DOI: 10.1124/pr.111.005439] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Metabolism represents the most prevalent mechanism for drug clearance. Many drugs are converted to metabolites that can retain the intrinsic affinity of the parent drug for the pharmacological target. Drug metabolism redox reactions such as heteroatom dealkylations, hydroxylations, heteroatom oxygenations, reductions, and dehydrogenations can yield active metabolites, and in rare cases even conjugation reactions can yield an active metabolite. To understand the contribution of an active metabolite to efficacy relative to the contribution of the parent drug, the target affinity, functional activity, plasma protein binding, membrane permeability, and pharmacokinetics of the active metabolite and parent drug must be known. Underlying pharmacokinetic principles and clearance concepts are used to describe the dispositional behavior of metabolites in vivo. A method to rapidly identify active metabolites in drug research is described. Finally, over 100 examples of drugs with active metabolites are discussed with regard to the importance of the metabolite(s) in efficacy and safety.
Collapse
Affiliation(s)
- R Scott Obach
- Pfizer Inc., Eastern Point Rd., Groton, CT 06340, USA.
| |
Collapse
|
124
|
Wright DFB, Stamp LK, Merriman TR, Barclay ML, Duffull SB, Holford NHG. The population pharmacokinetics of allopurinol and oxypurinol in patients with gout. Eur J Clin Pharmacol 2013; 69:1411-21. [DOI: 10.1007/s00228-013-1478-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 02/02/2013] [Indexed: 11/28/2022]
|
125
|
Stocker SL, McLachlan AJ, Savic RM, Kirkpatrick CM, Graham GG, Williams KM, Day RO. The pharmacokinetics of oxypurinol in people with gout. Br J Clin Pharmacol 2013; 74:477-89. [PMID: 22300439 DOI: 10.1111/j.1365-2125.2012.04207.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
AIMS Our aim was to identify and quantify the sources of variability in oxypurinol pharmacokinetics and explore relationships with plasma urate concentrations. METHODS Non-linear mixed effects modelling was applied to concentration-time data from 155 gouty patients with demographic, medical history and renal transporter genotype information. RESULTS A one compartment pharmacokinetic model with first order absorption best described the oxypurinol concentration-time data. Renal function and concomitant medicines (diuretics and probenecid), but not transporter genotype, significantly influenced oxypurinol pharmacokinetics and reduced the between subject variability in the apparent clearance of oxypurinol (CL/F(m)) from 65% to 29%. CL/F(m) for patients with normal, mild, moderate and severe renal impairment was 1.8, 0.6, 0.3 and 0.18 l h(-1), respectively. Model predictions showed a relationship between plasma oxypurinol and urate concentrations and failure to reach target oxypurinol concentrations using suggested allopurinol dosing guidelines. CONCLUSIONS In conclusion, this first established pharmacokinetic model provides a tool to achieve target oxypurinol plasma concentrations, thereby optimizing the effectiveness and safety of allopurinol therapy in gouty patients with various degrees of renal impairment.
Collapse
|
126
|
Jandová K, Riljak V, Pokorný J, Marešová D. Influence of allopurinol on evoked cortical afterdischarges during early ontogenesis. Physiol Res 2012; 61:S111-7. [PMID: 23130896 DOI: 10.33549/physiolres.932404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The aim of our study was to test the hypothesis, whether repeated allopurinol pre-treatment (in dose of 135 mg/kg s.c.) can influence changes of brain excitability caused by long-term hypoxia exposition in young immature rats. Rat pups were exposed together with their mother in to an intermittent hypobaric hypoxia (simulated altitude of 7 000 m) since the day of birth till the 11th day (youngest experimental group) or 17th day for 8 hours a day. Allopurinol was administered daily immediately before each hypoxia exposition. The duration of evoked afterdischarges (ADs) and the shape of evoked graphoelements were evaluated in 12, 18, 25 and 35-day-old freely moving male pups. Hypobaric hypoxia prolonged the duration of ADs in 12, 18 and 25-day-old rats. The ADs were prolonged in 35-day-old rats only after the first stimulation. Allopurinol shorted the duration of ADs only in 12-day-old pups. In older experimental group the effect of allopurinol treatment was less pronounced.
Collapse
Affiliation(s)
- K Jandová
- Institute of Physiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| | | | | | | |
Collapse
|
127
|
Paniz L, Schmidt AP, Souza DO. The modulatory effects of allopurinol onN-methyld-aspartate receptors in the central nervous system. Cell Biochem Funct 2012; 30:709-10. [DOI: 10.1002/cbf.2896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 08/24/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Lucas Paniz
- Department of Biochemistry, ICBS; Universidade Federal do Rio Grande do Sul; Porto Alegre; RS; Brazil
| | | | - Diogo O. Souza
- Department of Biochemistry, ICBS; Universidade Federal do Rio Grande do Sul; Porto Alegre; RS; Brazil
| |
Collapse
|
128
|
Curiel RV, Guzman NJ. Challenges Associated with the Management of Gouty Arthritis in Patients with Chronic Kidney Disease: A Systematic Review. Semin Arthritis Rheum 2012; 42:166-78. [DOI: 10.1016/j.semarthrit.2012.03.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 03/13/2012] [Accepted: 03/18/2012] [Indexed: 02/07/2023]
|
129
|
Vaughan VC, Sullivan-Gunn M, Hinch E, Martin P, Lewandowski PA. Eicosapentaenoic acid and oxypurinol in the treatment of muscle wasting in a mouse model of cancer cachexia. PLoS One 2012; 7:e45900. [PMID: 23029301 PMCID: PMC3447801 DOI: 10.1371/journal.pone.0045900] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/27/2012] [Indexed: 01/02/2023] Open
Abstract
Cancer cachexia is a wasting condition, driven by systemic inflammation and oxidative stress. This study investigated eicosapentaenoic acid (EPA) in combination with oxypurinol as a treatment in a mouse model of cancer cachexia. Mice with cancer cachexia were randomized into 4 treatment groups (EPA (0.4 g/kg/day), oxypurinol (1 mmol/L ad-lib), combination, or control), and euthanized after 29 days. Analysis of oxidative damage to DNA, mRNA analysis of pro-oxidant, antioxidant and proteolytic pathway components, along with enzyme activity of pro- and antioxidants were completed on gastrocnemius muscle. The control group displayed earlier onset of tumor compared to EPA and oxypurinol groups (P<0.001). The EPA group maintained body weight for an extended duration (20 days) compared to the oxypurinol (5 days) and combination (8 days) groups (P<0.05). EPA (18.2±3.2 pg/ml) and combination (18.4±3.7 pg/ml) groups had significantly higher 8-OH-dG levels than the control group (12.9±1.4 pg/ml, P≤0.05) indicating increased oxidative damage to DNA. mRNA levels of GPx1, MURF1 and MAFbx were higher following EPA treatment compared to control (P≤0.05). Whereas oxypurinol was associated with higher GPx1, MnSOD, CAT, XDH, MURF1, MAFbx and UbB mRNA compared to control (P≤0.05). Activity of total SOD was higher in the oxypurinol group (32.2±1.5 U/ml) compared to control (27.0±1.3 U/ml, P<0.01), GPx activity was lower in the EPA group (8.76±2.0 U/ml) compared to control (14.0±1.9 U/ml, P<0.05), and catalase activity was lower in the combination group (14.4±2.8 U/ml) compared to control (20.9±2.0 U/ml, P<0.01). There was no change in XO activity. The increased rate of weight decline in mice treated with oxypurinol indicates that XO may play a protective role during the progression of cancer cachexia, and its inhibition is detrimental to outcomes. In combination with EPA, there was little significant improvement from control, indicating oxypurinol is unlikely to be a viable treatment compound in cancer cachexia.
Collapse
MESH Headings
- Adenocarcinoma/complications
- Adenocarcinoma/drug therapy
- Animals
- Cachexia/drug therapy
- Cachexia/etiology
- Catalase/metabolism
- Cell Line, Tumor
- Drug Evaluation, Preclinical
- Drug Therapy, Combination
- Eicosapentaenoic Acid/pharmacology
- Eicosapentaenoic Acid/therapeutic use
- Enzyme Inhibitors/pharmacology
- Enzyme Inhibitors/therapeutic use
- Female
- Gene Expression/drug effects
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy/drug therapy
- Muscular Atrophy/etiology
- Neoplasm Transplantation
- Neoplasms, Experimental/complications
- Neoplasms, Experimental/drug therapy
- Organ Size/drug effects
- Oxidative Stress
- Oxypurinol/pharmacology
- Oxypurinol/therapeutic use
- Superoxide Dismutase/metabolism
- Tumor Burden
- Weight Loss/drug effects
- Xanthine Oxidase/antagonists & inhibitors
- Xanthine Oxidase/metabolism
Collapse
Affiliation(s)
| | | | - Edward Hinch
- School of Medicine, Deakin University, Waurn Ponds, Australia
| | - Peter Martin
- School of Medicine, Deakin University, Waurn Ponds, Australia
| | | |
Collapse
|
130
|
Day RO, Kannangara DRW, Hayes JM, Furlong TJ. Successful use of allopurinol in a patient on dialysis. BMJ Case Rep 2012; 2012:bcr.02.2012.5814. [PMID: 22675142 DOI: 10.1136/bcr.02.2012.5814] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
We report the case of a man with chronic tophaceous gout who had end-stage renal failure secondary to the Alport syndrome. Following a failed kidney transplant, where urate deposition was a suspected contributor, the patient responded positively to consistent allopurinol therapy and regular haemodialysis sessions. Extensive and destructive tophi receded in size remarkably and the almost constant incidence of acute attacks of gout subsided. The patient has recently received a new kidney transplant and his plasma concentrations of urate are controlled well with allopurinol and he no longer experiences acute attacks of gout. While efficacious, adherence is critical for achieving the therapeutic effects of allopurinol even in end-stage renal disease.
Collapse
Affiliation(s)
- Richard Osborne Day
- Department of Clinical Pharmacology & Toxicology, St Vincent's Hospital, Sydney, New South Wales, Australia.
| | | | | | | |
Collapse
|
131
|
Chiu MLS, Hu M, Ng MHL, Yeung CK, Chan JCY, Chang MM, Cheng SH, Li L, Tomlinson B. Association between HLA-B*58:01 allele and severe cutaneous adverse reactions with allopurinol in Han Chinese in Hong Kong. Br J Dermatol 2012; 167:44-9. [PMID: 22348415 DOI: 10.1111/j.1365-2133.2012.10894.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Allopurinol has been reported as a common cause of severe cutaneous adverse reactions (SCARs). Recent studies in various populations suggest that HLA-B*58:01 is a strong genetic marker for allopurinol-induced SCAR, especially in populations with a high frequency of HLA-B*58:01. OBJECTIVES To confirm the association link between HLA-B*58:01 and hypersensitivity reactions attributed to allopurinol use in Han Chinese patients in Hong Kong. METHODS We performed a case-control study to investigate whether the HLA-B*58:01 allele predisposes to allopurinol-induced SCAR in Han Chinese patients in Hong Kong. The HLA-B*58:01 genotyping was performed in 20 patients with allopurinol-induced SCAR or erythema multiforme major (EMM; n = 1) and in 30 patients tolerant to allopurinol. RESULTS All of the 19 patients with allopurinol-induced SCAR examined but not the patient with EMM carried HLA-B*58:01 whereas only four (13%) of the control patients had this allele. The positive rate of the HLA-B*58:01 was significantly higher in the cases than in the allopurinol-tolerant control group [odds ratio (OR) 123·5, 95% confidence interval (CI) 12·8-1195·1; P < 1 × 10(-4) ] and was even higher after removal of the patient with EMM (OR 229·7, 95% CI 11·7-4520·4). The sensitivity and specificity of the HLA-B*58:01 allele for prediction of allopurinol-induced SCAR were 100% and 86·7%, respectively. CONCLUSIONS This study confirmed the strong association between the HLA-B*58:01 and allopurinol-induced SCAR in Hong Kong Han Chinese patients. A screening test for the HLA-B*58:01 allele should effectively reduce the risk for allopurinol-induced SCAR in Chinese populations.
Collapse
Affiliation(s)
- M L S Chiu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Kannangara DRW, Roberts DM, Furlong TJ, Graham GG, Williams KM, Day RO. Oxypurinol, allopurinol and allopurinol-1-riboside in plasma following an acute overdose of allopurinol in a patient with advanced chronic kidney disease. Br J Clin Pharmacol 2011; 73:828-9. [PMID: 22098112 DOI: 10.1111/j.1365-2125.2011.04147.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
133
|
Smith HS, Bracken D, Smith JM. Gout: Current Insights and Future Perspectives. THE JOURNAL OF PAIN 2011; 12:1113-29. [DOI: 10.1016/j.jpain.2011.06.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 06/15/2011] [Accepted: 06/24/2011] [Indexed: 02/07/2023]
|
134
|
Frezzatti R, Silveira PF. Allopurinol reduces the lethality associated with acute renal failure induced by Crotalus durissus terrificus snake venom: comparison with probenecid. PLoS Negl Trop Dis 2011; 5:e1312. [PMID: 21909449 PMCID: PMC3167791 DOI: 10.1371/journal.pntd.0001312] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 07/27/2011] [Indexed: 02/07/2023] Open
Abstract
Background Acute renal failure is one of the most serious complications of envenoming resulting from Crotalus durissus terrificus bites. This study evaluated the relevance of hyperuricemia and oxidative stress and the effects of allopurinol and probenecid in renal dysfunction caused by direct nephrotoxicity of C. d. terrificus venom. Methodology/Principal Findings Hematocrit, protein, renal function and redox status were assessed in mice. High ratio of oxidized/reduced glutathione and hyperuricemia induced by C. d. terrificus venom were ameliorated by both, allopurinol or probenecid, but only allopurinol significantly reduced the lethality caused by C. d. terrificus venom. The effectiveness of probenecid is compromised probably because it promoted hypercreatinemia and hypocreatinuria and worsed the urinary hypo-osmolality in envenomed mice. In turn, the highest effectiveness of allopurinol might be due to its ability to diminish the intracellular formation of uric acid. Conclusions/Significance Data provide consistent evidences linking uric acid with the acute renal failure induced by C. d. terrificus venom, as well as that this envenoming in mice constitutes an attractive animal model suitable for studying the hyperuricemia and that the allopurinol deserves to be clinically evaluated as an approach complementary to anti-snake venom serotherapy. In Brazil, among registered snake bites, those by the genus Crotalus originate the highest mortality rate. The rattlesnake Crotalus durissus terrificus is the most frequently implicated in these accidents. The kidney is a particularly vulnerable organ to the venom of this rattlesnake. In fact, the most serious complication of Crotalus snake bite is the renal dysfunction, and among the fatal cases of Crotalus bites in Brazil 5% are patients treated with antivenom. The hyperuricemia has been observed in human accidents with snake venoms, but this parameter has not received any special attention as a relevant factor in the etiology of renal dysfunction caused by these venoms. This study examined the effects of treatments with low-cost and low-risk uricostatic (allopurinol) and uricosuric (probenecid) drugs on the envenomation by C. d. terrificus, showing that allopurinol and probenecid mitigated certain nephrotoxic effects, as well as the survival of envenomed mice was improved through the effects of allopurinol on reduction of oxidative stress and intracellular formation of uric acid. This new knowledge provides consistent evidences linking uric acid with the renal dysfunction induced by rattlesnake bites and that the allopurinol deserves to be clinically evaluated as an approach complementary to anti-snake venom serotherapy.
Collapse
Affiliation(s)
- Rodrigo Frezzatti
- Laboratory of Pharmacology, Instituto Butantan, São Paulo, São Paulo, Brazil
| | - Paulo Flavio Silveira
- Laboratory of Pharmacology, Instituto Butantan, São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
135
|
Graham GG, Punt J, Arora M, Day RO, Doogue MP, Duong JK, Furlong TJ, Greenfield JR, Greenup LC, Kirkpatrick CM, Ray JE, Timmins P, Williams KM. Clinical pharmacokinetics of metformin. Clin Pharmacokinet 2011; 50:81-98. [PMID: 21241070 DOI: 10.2165/11534750-000000000-00000] [Citation(s) in RCA: 829] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metformin is widely used for the treatment of type 2 diabetes mellitus. It is a biguanide developed from galegine, a guanidine derivative found in Galega officinalis (French lilac). Chemically, it is a hydrophilic base which exists at physiological pH as the cationic species (>99.9%). Consequently, its passive diffusion through cell membranes should be very limited. The mean ± SD fractional oral bioavailability (F) of metformin is 55 ± 16%. It is absorbed predominately from the small intestine. Metformin is excreted unchanged in urine. The elimination half-life (t(½)) of metformin during multiple dosages in patients with good renal function is approximately 5 hours. From published data on the pharmacokinetics of metformin, the population mean of its clearances were calculated. The population mean renal clearance (CL(R)) and apparent total clearance after oral administration (CL/F) of metformin were estimated to be 510 ± 130 mL/min and 1140 ± 330 mL/min, respectively, in healthy subjects and diabetic patients with good renal function. Over a range of renal function, the population mean values of CL(R) and CL/F of metformin are 4.3 ± 1.5 and 10.7 ± 3.5 times as great, respectively, as the clearance of creatinine (CL(CR)). As the CL(R) and CL/F decrease approximately in proportion to CL(CR), the dosage of metformin should be reduced in patients with renal impairment in proportion to the reduced CL(CR). The oral absorption, hepatic uptake and renal excretion of metformin are mediated very largely by organic cation transporters (OCTs). An intron variant of OCT1 (single nucleotide polymorphism [SNP] rs622342) has been associated with a decreased effect on blood glucose in heterozygotes and a lack of effect of metformin on plasma glucose in homozygotes. An intron variant of multidrug and toxin extrusion transporter [MATE1] (G>A, SNP rs2289669) has also been associated with a small increase in antihyperglycaemic effect of metformin. Overall, the effect of structural variants of OCTs and other cation transporters on the pharmacokinetics of metformin appears small and the subsequent effects on clinical response are also limited. However, intersubject differences in the levels of expression of OCT1 and OCT3 in the liver are very large and may contribute more to the variations in the hepatic uptake and clinical effect of metformin. Lactic acidosis is the feared adverse effect of the biguanide drugs but its incidence is very low in patients treated with metformin. We suggest that the mean plasma concentrations of metformin over a dosage interval be maintained below 2.5 mg/L in order to minimize the development of this adverse effect.
Collapse
Affiliation(s)
- Garry G Graham
- Department of Pharmacology & Toxicology, St Vincents Clinical School, University of New South Wales, Sydney, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Relationship Between Serum Urate and Plasma Oxypurinol in the Management of Gout: Determination of Minimum Plasma Oxypurinol Concentration to Achieve a Target Serum Urate Level. Clin Pharmacol Ther 2011; 90:392-8. [DOI: 10.1038/clpt.2011.113] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
137
|
Pandya BJ, Riedel AA, Swindle JP, Becker LK, Hariri A, Dabbous O, Krishnan E. Relationship between physician specialty and allopurinol prescribing patterns: a study of patients with gout in managed care settings. Curr Med Res Opin 2011; 27:737-44. [PMID: 21271794 DOI: 10.1185/03007995.2011.552570] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Allopurinol is used to lower serum uric acid (sUA) levels in gout patients. The objective of this study was to investigate the influence of physician specialty on allopurinol treatment patterns and sUA control. DESIGN AND METHODS This was a retrospective study using claims from a managed care database of US health plan enrollees. Gout patients at least 18 years of age who received allopurinol were identified from the database between January 1, 2002 and April 30, 2007. The index date was defined as the date of the earliest allopurinol claim, and patients were required to have health plan enrollment for at least 365 days prior to and following the index date for inclusion. Physician specialty was determined using the index allopurinol claim. Dosage of allopurinol prescription(s) and number of gout flares were determined from claims data. sUA measurements were used to assess goal attainment over a period of at least one year following the index allopurinol prescription. RESULTS There were 3363 patients with gout of whom 69.9% received an index allopurinol prescription from a generalist/internist, 5.7% from a rheumatologist, 2.6% from a nephrologist, and 21.8% from a physician with other specialty. Of patients receiving their index prescription from a nephrologist, 38.7% reached the sUA goal of <6 mg/dL (357 μmol/L), as compared to patients prescribed by a rheumatologist, generalist/internist, or other physician (35.4%, 31.4%, and 39.4%, respectively; P = 0.015). When controlling for patient characteristics, multivariate analysis did not reveal statistically significant different odds of sUA goal attainment based on prescribing physician specialty, though separate analyses indicated that patients prescribed by a nephrologist had fewer gout flares. Change in allopurinol dosage from initial to final dose was more frequent among patients prescribed by rheumatologists and nephrologists. CONCLUSION There is significant heterogeneity in the specialists' management of sUA levels in patients with gout, possibly reflecting differences in case mix and treatment approaches. Limitations related to the use of claims data, such as inability to observe medications filled over-the-counter, should be considered when interpreting study results.
Collapse
Affiliation(s)
- Bhavik J Pandya
- Takeda Pharmaceutical International, Inc., Deerfield, IL, USA.
| | | | | | | | | | | | | |
Collapse
|
138
|
STOCKER SOPHIEL, GRAHAM GARRYG, McLACHLAN ANDREWJ, WILLIAMS KENNETHM, DAY RICHARDO. Pharmacokinetic and Pharmacodynamic Interaction Between Allopurinol and Probenecid in Patients with Gout. J Rheumatol 2011; 38:904-10. [DOI: 10.3899/jrheum.101160] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Objective.To investigate the pharmacokinetic and pharmacodynamic interaction between probenecid and oxypurinol (the active metabolite of allopurinol) in patients with gout.Methods.This was an open-label observational clinical study. Blood and urine samples were collected to measure oxypurinol and urate concentrations. We examined the effects of adding probenecid to allopurinol therapy upon plasma concentrations and renal clearances of urate and oxypurinol.Results.Twenty patients taking allopurinol 100–400 mg daily completed the study. Maximum coadministered doses of probenecid were 250 mg/day (n = 1), 500 mg/day (n = 19), 1000 mg/day (n = 7), 1500 mg/day (n = 3), and 2000 mg/day (n = 1). All doses except the 250 mg daily dose were divided and dosing was twice daily. Estimated creatinine clearances ranged from 28 to 113 ml/min. Addition of probenecid 500 mg/day to allopurinol therapy decreased plasma urate concentrations by 25%, from mean 0.37 mmol/l (95% CI 0.33–0.41) to mean 0.28 mmol/l (95% CI 0.24–0.32) (p < 0.001); and increased renal urate clearance by 62%, from mean 6.0 ml/min (95% CI 4.5–7.5) to mean 9.6 ml/min (95% CI 6.9–12.3) (p < 0.001). Average steady-state plasma oxypurinol concentrations decreased by 26%, from mean 11.1 mg/l (95% CI 5.0–17.3) to mean 8.2 mg/l (95% CI 4.0–12.4) (p < 0.001); and renal oxypurinol clearance increased by 24%, from mean 12.7 ml/min (95% CI 9.6–15.8) to mean 16.1 ml/min (95% CI 12.0–20.2) (p < 0.05). The additional hypouricemic effect of probenecid 500 mg/day appeared to be lower in patients with renal impairment.Conclusion.Coadministration of allopurinol with probenecid had a significantly greater hypouricemic effect than allopurinol alone despite an associated reduction of plasma oxypurinol concentrations. Australian Clinical Trials Registry ACTRN012606000276550.
Collapse
|
139
|
Ng DY, Stocker SL, Graham GG, Williams KM, Day RO. Lack of effect of hydrochlorothiazide and low-dose aspirin on the renal clearance of urate and oxypurinol after a single dose of allopurinol in normal volunteers. Eur J Clin Pharmacol 2010; 67:709-13. [PMID: 21181139 DOI: 10.1007/s00228-010-0963-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 11/23/2010] [Indexed: 11/30/2022]
Abstract
AIMS To determine whether low-dose aspirin and hydrochlorothiazide (HCTZ) affect the renal clearance of oxypurinol and/or urate. METHODS Healthy volunteers (n = 8) were treated with allopurinol (600 mg, control), and allopurinol (600 mg) co-administered with single doses of aspirin (100 mg) or HCTZ (25 mg) or a combination of the two. RESULTS Hydrochlorothiazide, low-dose aspirin or a combination of the two, when co-administered with allopurinol, did not significantly alter (P > 0.05) the renal clearance of oxypurinol or urate. In particular, aspirin and HCTZ, when taken together and with allopurinol, did not change (P > 0.05) oxypurinol fractional renal clearance (allopurinol alone: 0.217, 0.173-0.262; combined: 0.202, 0.155-0.250) or urate fractional renal clearance (allopurinol alone: 0.066, 0.032-0.099; combined: 0.058, 0.038-0.078). CONCLUSIONS A single, low-dose of aspirin or an anti-hypertensive dose of hydrochlorothiazide, when administered alone or together with allopurinol, are unlikely to alter the hypouricaemic effect of allopurinol. The effect of chronic aspirin and HCTZ dosing taken together upon the efficacy of chronic allopurinol therapy in patients with hyperuricaemia needs to be investigated.
Collapse
Affiliation(s)
- Daniel Y Ng
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Xavier Level 2, Darlinghurst, NSW 2010, Australia
| | | | | | | | | |
Collapse
|
140
|
Abstract
For decades allopurinol has been used as a xanthine oxidase inhibitor for treatment of hyperuricemia and gout. Although effective in many patients, some experience sensitivity to the drug. In some cases, this sensitivity may lead to allopurinol hypersensitivity disorder, which if untreated can be fatal. Recently the Food and Drug Administration has approved the use of febuxostat as an alternative therapy for hyperuricemia and gout. Febuxostat is a new xanthine oxidase inhibitor, but is not purine based and therefore decreases adverse reactions due to patient sensitivity. This review is a comprehensive look at the background of hyperuricemia and gout treatment with allopurinol compared to recent clinical studies with febuxostat. Each clinical study is evaluated and summarized, identifying the advances in treatment that have been made as well as the concerns that still exist with either treatment.
Collapse
Affiliation(s)
- Amy L. Stockert
- Ohio Northern University, The Raabe College of Pharmacy, 525 N. Main St. Ada, OH 45810, USA
| | - Melissa Stechschulte
- Ohio Northern University, The Raabe College of Pharmacy, 525 N. Main St. Ada, OH 45810, USA
| |
Collapse
|
141
|
Measurement of urinary oxypurinol by high performance liquid chromatography–tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2010; 878:2363-8. [DOI: 10.1016/j.jchromb.2010.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 07/14/2010] [Accepted: 07/20/2010] [Indexed: 11/17/2022]
|
142
|
|
143
|
Taibi G, Carruba G, Miceli V, Cocciadiferro L, Cucchiara A, Nicotra CMA. Sildenafil protects epithelial cell through the inhibition of xanthine oxidase and the impairment of ROS production. Free Radic Res 2010; 44:232-239. [PMID: 19968586 DOI: 10.3109/10715760903431426] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Xanthine oxidase (XO) plays an important role in various forms of ischemic and vascular injuries, inflammatory diseases and chronic heart failure. The XO inhibitors allopurinol and oxypurinol held considerable promise in the treatment of these conditions both in experimental animals and in human clinical trials. More recently, an endothelium-based protective effect of sildenafil has been reported in preconditioning prior to ischemia/reperfusion in healthy human subjects. Based on the structural similarities between allopurinol and oxypurinol with sildenafil and with zaprinast the authors have investigated the potential effects of these latter compounds on the buttermilk XO and on non-tumourigenic (HMEC) and malignant (MCF7) human mammary epithelial cells. Both sildenafil and zaprinast induced a significant and consistent decrease of XO expression and activity in either cell line. In MCF7 cells only, this effect was associated with the abrogation of xanthine-induced cytotoxicity. Overall, the data suggest that the protective effect of sildenafil on epithelial cells is a consequence of the inhibition of the XO and of the resulting decrease of free oxygen radical production that may influence the expression of NADPH oxidase and PDE-5.
Collapse
Affiliation(s)
- Gennaro Taibi
- Dipartimento di Oncologia Sperimentale e Applicazioni Cliniche, Sezione di Biochimica e Oncologia Clinica, AOUP Paolo Giaccone', Università di Palermo, Via Del Vespro 129, 90127 Palermo, Italy.
| | | | | | | | | | | |
Collapse
|
144
|
|
145
|
Pascual E, Sivera F. Gout: new advances in the diagnosis and management of an old disease. ACTA ACUST UNITED AC 2009. [DOI: 10.2217/ijr.09.5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
146
|
van Dijk AJ, Parvizi N, Taverne MAM, Fink-Gremmels J. Placental transfer and pharmacokinetics of allopurinol in late pregnant sows and their fetuses. J Vet Pharmacol Ther 2009; 31:489-95. [PMID: 19000269 DOI: 10.1111/j.1365-2885.2008.00976.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
At present no standard pharmacological intervention strategy is available to reduce these adverse effects of birth asphyxia. In the present study we aimed to evaluate placental transfer of allopurinol, an inhibitor of XOR. For this purpose, fetal catheterization of the jugular vein was conducted in five late pregnant sows (one fetus per sow). At 24-48 h after surgery, sows received allopurinol (15 mg/kg body weight; i.v.) and pharmacokinetics of allopurinol and its active metabolite oxypurinol were measured in both late pregnant sows and fetuses. Maternal and fetal blood samples were collected during and after allopurinol administration. Maternal C(max) values averaged 41.90 microg/mL (allopurinol) and 3.68 microg/mL (oxypurinol). Allopurinol crossed the placental barrier as shown by the average fetal C(max) values of 5.05 microg/mL at 1.47 h after allopurinol administration to the sow. In only one fetus low plasma oxypurinol concentrations were found. Incubations of subcellular hepatic fractions of sows and 24-h-old piglets confirmed that allopurinol could be metabolized into oxypurinol. In conclusion, we demonstrated that allopurinol can cross the placental barrier, a prerequisite for further studies evaluating the use of allopurinol as a neuroprotective agent to reduce the adverse effects following birth asphyxia in neonatal piglets.
Collapse
Affiliation(s)
- A J van Dijk
- Department of Veterinary Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
147
|
Reinders MK, van Roon EN, Jansen TLTA, Delsing J, Griep EN, Hoekstra M, van de Laar MAFJ, Brouwers JRBJ. Efficacy and tolerability of urate-lowering drugs in gout: a randomised controlled trial of benzbromarone versus probenecid after failure of allopurinol. Ann Rheum Dis 2009; 68:51-6. [PMID: 18250112 DOI: 10.1136/ard.2007.083071] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To investigate the efficacy and tolerability of allopurinol as the first-choice antihyperuricaemic treatment for gout, and compare the efficacy and tolerability of benzbromarone and probenecid as second-choice treatment. METHODS Prospective, multicentre, open-label, two-stage randomised controlled trial in gout patients with normal renal function. Enrolled patients were given 300 mg allopurinol for 2 months (stage 1). Those patients who could not tolerate allopurinol or who did not attain the target serum urate concentration (sUr) < or=0.30 mmol/l (5.0 mg/dl), which was defined as successful, were randomised to benzbromarone 200 mg/day or probenecid 2 g/day for another 2 months (stage 2). RESULTS 96 patients were enrolled in stage 1. 82 patients (85%) were eligible for the analysis at the end of stage 1: there was a mean (SD) decrease in sUr concentration of 35 (11)% from baseline; 20 patients (24%) attained target sUr < or=0.30 mmol/l; and 9 patients (11%) stopped allopurinol because of adverse drug reactions. 62 patients were enrolled in stage 2. 27 patients received benzbromarone (3 patients not eligible for analysis) and 35 received probenecid (4 patients not eligible for analysis). Treatment with benzbromarone was successful in 22/24 patients (92%) and with probenecid in 20/31 patients (65%) (p = 0.03 compared with benzbromarone). Compared with baseline values, there was a mean (SD) decrease of sUr concentration of 64 (9)% with benzbromarone and 50 (7)% with probenecid (p<0.001). CONCLUSION This study showed that allopurinol 300 mg/day has a poor efficacy and tolerability profile when used to attain a biochemical predefined target level of sUr < or =0.30 mmol/l, following 2 months of treatment. In stage 2, benzbromarone 200 mg/day was more effective and better tolerated than probenecid 2 g/day.
Collapse
Affiliation(s)
- M K Reinders
- Department of Clinical Pharmacy and Pharmacology, Medisch Centrum Leeuwarden, Leeuwarden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Schmidt AP, Böhmer AE, Antunes C, Schallenberger C, Porciúncula LO, Elisabetsky E, Lara DR, Souza DO. Anti-nociceptive properties of the xanthine oxidase inhibitor allopurinol in mice: role of A1 adenosine receptors. Br J Pharmacol 2009; 156:163-72. [PMID: 19133997 PMCID: PMC2697763 DOI: 10.1111/j.1476-5381.2008.00025.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 07/25/2008] [Accepted: 09/02/2008] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Allopurinol is a potent inhibitor of the enzyme xanthine oxidase, used primarily in the treatment of hyperuricemia and gout. It is well known that purines exert multiple effects on pain transmission. We hypothesized that the inhibition of xanthine oxidase by allopurinol, thereby reducing purine degradation, could be a valid strategy to enhance purinergic activity. The aim of this study was to investigate the anti-nociceptive profile of allopurinol on chemical and thermal pain models in mice. EXPERIMENTAL APPROACH Mice received an intraperitoneal (i.p.) injection of vehicle (Tween 10%) or allopurinol (10-400 mg kg(-1)). Anti-nociceptive effects were measured with intraplantar capsaicin, intraplantar glutamate, tail-flick or hot-plate tests. KEY RESULTS Allopurinol presented dose-dependent anti-nociceptive effects in all models. The opioid antagonist naloxone did not affect these anti-nociceptive effects. The non-selective adenosine-receptor antagonist caffeine and the selective A(1) adenosine-receptor antagonist, DPCPX, but not the selective A(2A) adenosine-receptor antagonist, SCH58261, completely prevented allopurinol-induced anti-nociception. No obvious motor deficits were produced by allopurinol, at doses up to 200 mg kg(-1). Allopurinol also caused an increase in cerebrospinal fluid levels of purines, including the nucleosides adenosine and guanosine, and decreased cerebrospinal fluid concentration of uric acid. CONCLUSIONS AND IMPLICATIONS Allopurinol-induced anti-nociception may be related to adenosine accumulation. Allopurinol is an old and extensively used compound and seems to be well tolerated with no obvious central nervous system toxic effects at high doses. This drug may be useful to treat pain syndromes in humans.
Collapse
Affiliation(s)
- A P Schmidt
- Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Hanlon JT, Aspinall SL, Semla TP, Weisbord SD, Fried LF, Good CB, Fine MJ, Stone RA, Pugh MJV, Rossi MI, Handler SM. Consensus guidelines for oral dosing of primarily renally cleared medications in older adults. J Am Geriatr Soc 2008; 57:335-40. [PMID: 19170784 DOI: 10.1111/j.1532-5415.2008.02098.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OBJECTIVES To establish consensus oral dosing guidelines for primarily renally cleared medications prescribed for older adults. DESIGN Literature search followed by a two-round modified Delphi survey. SETTING A nationally representative survey of experts in geriatric clinical pharmacy. PARTICIPANTS Eleven geriatric clinical pharmacists. MEASUREMENTS After a comprehensive literature search and review by an investigative group of six physicians (2 general internal medicine, 2 nephrology, 2 geriatrics), 43 dosing recommendations for 30 medications at various levels of renal function were created. The expert panel rated its agreement with each of these 43 dosing recommendations using a 5-point Likert scale (1=strongly disagree to 5=strongly agree). Recommendation-specific means and 95% confidence intervals were estimated. Consensus was defined as a lower 95% confidence limit of greater than 4.0 for the recommendation-specific mean score. RESULTS The response rate was 81.8% (9/11) for the first round. All respondents who completed the first round also completed the second round. The expert panel reached consensus on 26 recommendations involving 18 (60%) medications. For 10 medications (chlorpropamide, colchicine, cotrimoxazole, glyburide, meperidine, nitrofurantoin, probenecid, propoxyphene, spironolactone, and triamterene), the consensus recommendation was not to use the medication in older adults below a specified level of renal function (e.g., creatinine clearance <30 mL/min). For the remaining eight medications (acyclovir, amantadine, ciprofloxacin, gabapentin, memantine, ranitidine, rimantadine, and valacyclovir), specific recommendations for dose reduction or interval extension were made. CONCLUSION An expert panel of geriatric clinical pharmacists was able to reach consensus agreement on a number of oral medications that are primarily renally cleared.
Collapse
Affiliation(s)
- Joseph T Hanlon
- Department of Medicine, University of Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Stocker SL, Williams KM, McLachlan AJ, Graham GG, Day RO. Pharmacokinetic and pharmacodynamic interaction between allopurinol and probenecid in healthy subjects. Clin Pharmacokinet 2008; 47:111-8. [PMID: 18193917 DOI: 10.2165/00003088-200847020-00004] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND AND OBJECTIVE Combination therapy with allopurinol and probenecid is used to treat tophaceous gout in patients who do not respond sufficiently to allopurinol alone. However, the potential interaction between these drugs has not been systematically investigated. The objective of this study was to investigate the pharmacokinetics and hypouricaemic effect of oxypurinol (the active metabolite of allopurinol) and probenecid when administered alone and in combination in healthy subjects. METHODS An open-label, randomized, three-way crossover clinical trial was conducted in 12 healthy adults. Subjects were randomized to receive treatment for 7 days with allopurinol (150 mg twice daily), probenecid (500 mg twice daily) or combination therapy with both drugs, with a 7-day washout period between treatments. Venous blood samples were collected predose (at 0 hours) and 1, 2, 3, 4, 6, 8, 10 and 12 hours after dosage for determination of oxypurinol and/or probenecid concentrations. Plasma and urinary urate concentrations were determined on each study day and at the end of each washout period. Pharmacokinetic and pharmacodynamic parameters were analysed using two-way ANOVA. RESULTS Coadministration of allopurinol and probenecid significantly reduced average steady-state plasma oxypurinol concentrations (mean+/-SD: allopurinol alone 9.7+/-2.1 mg/L vs combination 5.1+/-1.0 mg/L, p<0.001). Probenecid concentrations were unaffected. Plasma urate concentrations decreased (p<0.01) during allopurinol therapy (0.16+/-0.05 mmol/L), probenecid therapy (0.13+/-0.02 mmol/L) and combination therapy (0.09+/-0.02 mmol/L) compared with baseline (0.30+/-0.05 mmol/L). CONCLUSION Coadministration of allopurinol and probenecid to healthy subjects had a greater hypouricaemic effect than either allopurinol or probenecid alone, despite a reduction in plasma oxypurinol concentrations when the drugs were taken concomitantly.
Collapse
Affiliation(s)
- Sophie L Stocker
- Faculty of Pharmacy, University of Sydney, Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, New South Wales, Australia
| | | | | | | | | |
Collapse
|